US10696659B2 - Compounds having estrogen receptor alpha degradation activity and uses thereof - Google Patents

Compounds having estrogen receptor alpha degradation activity and uses thereof Download PDF

Info

Publication number
US10696659B2
US10696659B2 US16/690,611 US201916690611A US10696659B2 US 10696659 B2 US10696659 B2 US 10696659B2 US 201916690611 A US201916690611 A US 201916690611A US 10696659 B2 US10696659 B2 US 10696659B2
Authority
US
United States
Prior art keywords
hydroxyphenyl
phenylbut
dione
phenoxy
piperidine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US16/690,611
Other languages
English (en)
Other versions
US20200157078A1 (en
Inventor
Jie Fan
Ke Liu
Hui Zhang
Wei He
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Accutar Biotechnology Inc
Original Assignee
Accutar Biotechnology Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Accutar Biotechnology Inc filed Critical Accutar Biotechnology Inc
Priority to US16/690,611 priority Critical patent/US10696659B2/en
Priority to US16/851,994 priority patent/US11117885B2/en
Publication of US20200157078A1 publication Critical patent/US20200157078A1/en
Application granted granted Critical
Publication of US10696659B2 publication Critical patent/US10696659B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/08Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms
    • C07D211/18Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D211/30Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms with hydrocarbon radicals, substituted by doubly bound oxygen or sulfur atoms or by two oxygen or sulfur atoms singly bound to the same carbon atom
    • C07D211/32Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms with hydrocarbon radicals, substituted by doubly bound oxygen or sulfur atoms or by two oxygen or sulfur atoms singly bound to the same carbon atom by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C303/00Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides
    • C07C303/26Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides of esters of sulfonic acids
    • C07C303/28Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides of esters of sulfonic acids by reaction of hydroxy compounds with sulfonic acids or derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C309/00Sulfonic acids; Halides, esters, or anhydrides thereof
    • C07C309/63Esters of sulfonic acids
    • C07C309/64Esters of sulfonic acids having sulfur atoms of esterified sulfo groups bound to acyclic carbon atoms
    • C07C309/65Esters of sulfonic acids having sulfur atoms of esterified sulfo groups bound to acyclic carbon atoms of a saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/08Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms
    • C07D295/084Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/088Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D309/08Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D309/10Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/10Spiro-condensed systems

Definitions

  • the present disclosure relates to novel compounds having estrogen receptor alpha degradation activity, pharmaceutical compositions containing such compounds, and their use in prevention and treatment of diseases and conditions, e.g., cancer.
  • Estrogen a female sex hormone, through binding to its cognate Estrogen receptors, ER ⁇ and ER ⁇ , governs a wide range of physiological processes, e.g., the development of the female reproductive system, the maintenance of bone mass, and the protection of cardiovascular tissue and the central nervous system.
  • ER estrogen receptor
  • the receptor Upon estrogen's binding to an estrogen receptor (“ER”), the receptor undergoes a conformational change resulting in its homodimerization.
  • the ER homodimer then binds to estrogen-response elements (“EREs”) that are present in the promoters of a specific set of target genes and regulates their expression with the help of transcriptional coregulators.
  • EEEs estrogen-response elements
  • ER signaling is implicated in many pathways, it is well known that deregulation of ER signaling, specifically through ER ⁇ , results in uncontrolled cellular proliferation which eventually results into cancer.
  • ER+ breast cancer accounts for approximately 75% of all breast cancers diagnosed, as well as some ovarian and endometrial cancers. The prevalence of ER+ cancer has led to decades of investigation and development of antiestrogens as therapeutic agents.
  • Antiestrogen (i.e., hormonal) therapy is the first choice for treatment of most ER+ breast cancers.
  • antiestrogen therapies including aromatase inhibitors (e.g., letrozole and anastrozole); selective estrogen receptor modulators (e.g., tamoxifen, toremifene, and raloxifene); and selective estrogen receptor degraders (e.g., fulvestrant).
  • aromatase inhibitors e.g., letrozole and anastrozole
  • selective estrogen receptor modulators e.g., tamoxifen, toremifene, and raloxifene
  • selective estrogen receptor degraders e.g., fulvestrant
  • the aforementioned therapies may result in deleterious effects.
  • administration of aromatase inhibitors results in a decrease in bone mineral density, which can result in an increased risk of fractures.
  • Administration of selective estrogen modulators can result in development of endometrial cancer and/or cardiovascular issues, e.g., deep thrombosis and pulmonary embolism. Additionally, the aforementioned therapies may suffer from insufficient clinical efficacy.
  • ER ⁇ degradation may occur when both ER ⁇ and a ubiquitin ligase are bound and brought into close proximity.
  • E3 ubiquitin ligase is a ubiquitin ligase that CRBN forms an E3 ubiquitin ligase complex with damaged DNA binding protein 1 and Cullin 4. It functions as a substrate receptor by bringing the substrates to close proximity for ubiquitination and subsequent degradation by proteasomes.
  • small molecules drugs e.g., thalidomide and its close analogs, lenalidomide and pomalidomide, can simultaneously interact with CRBN and some other proteins. In doing so, CRBN may be exploited for target protein degradation, such as IKZF1 and IKZF3. This is thought to account for the anti-myeloma effects of thalidomide and related compounds.
  • provided herein are compounds of Formula (I), or a tautomer, stereoisomer or a mixture of stereoisomers, pharmaceutically acceptable salt, or hydrate thereof:
  • X 1 and X 2 are each independently selected from C(R 3 ) 2 , NR 4 , O, S, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 ;
  • A is selected from:
  • L* is a linker of 1 to 22 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • R 1 and R 2 are each independently selected from H, C 1 -C 6 alkyl, halo, alkoxy, acyloxy, hydroxy, and sulfhydryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • each R 3 is independently selected from H, C 1 -C 6 alkyl, halo, and hydroxy;
  • each R 5 is independently selected from C 1 -C 6 alkyl, halo, cyano, and hydroxy,
  • the compound of Formula (I) may encompass both the E and Z isomers. In some embodiments, the compound of Formula (I) may be a mixture of trans- and -cis olefin.
  • A may be any organic compound
  • A may be any organic compound
  • A may be any organic compound
  • A may be any organic compound
  • X 1 and X 2 are each independently selected from C(R 3 ) 2 , NR 4 , O, S, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 ;
  • B is selected from 5- to 6-member cycloalkyl, 5- to 6-member aryl, 5- to 6-member heterocycle, and 5- to 6-member heteroaryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • C is selected from:
  • L* is a linker of 1 to 22 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • R 1 and R 2 are each independently selected from H, C 1 -C 6 alkyl, halo, alkoxy, acyloxy, hydroxy, and sulfhydryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • each R 3 is independently selected from H, C 1 -C 6 alkyl, halo, and hydroxy;
  • each R 4 is independently selected from H, C 1 -C 6 alkyl, and acyl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • each R 5 is independently selected from C 1 -C 6 alkyl, halo, cyano, and hydroxy,
  • C may be
  • C may be
  • Also provided herein is a method of treating cancer in a subject in need thereof, comprising administering to said subject an effective amount of a compound disclosed herein.
  • the cancer is selected from breast cancer, lung cancer, ovarian cancer, endometrial cancer, prostate cancer, and esophageal cancer.
  • FIG. 1 illustrates an exemplary compound of the present disclosure bound to ER ⁇ and an E3 ubiquitin ligase cereblon.
  • FIG. 2A illustrates the ER ⁇ degradative activity of exemplary compound 15 of the present disclosure in a T47D cell line 4 hours after administration.
  • FIG. 2B illustrates the ER ⁇ degradative activity of exemplary compounds 54 and 64 of the present disclosure in a T47D cell line 6 hours after administration.
  • FIG. 2C illustrates the ER ⁇ degradative activity of exemplary compounds 54 and 64 of the present disclosure in a T47D cell line 6 hours after administration.
  • FIG. 2D illustrates the ER ⁇ degradative activity of exemplary compounds 8, 15, 54, 64, 88, 89, 108, 126, 143, and 120 of the present disclosure in a T47D cell line 8 hours after administration.
  • FIG. 2E illustrates the ER ⁇ degradative activity of exemplary compound 64 of the present disclosure at a concentration of 100 nM, as a function of time, in a T47D cell line.
  • FIG. 3A illustrates the ER ⁇ degradative activity of exemplary compounds 15, 54, and 64 of the present disclosure in an MCF7 cell line 4 hours after administration.
  • FIG. 3B illustrates the ER ⁇ degradative activity of exemplary compounds 54 and 64 of the present disclosure in an MCF7 cell line 6 hours after administration.
  • FIG. 3C illustrates the ER ⁇ degradative activity of exemplary compounds 160a, 184a, 17a, 16a, 31a, 28a, 32a, 29a, 161a, and 185a of the present disclosure in an MCF7 cell line 6 hours after administration.
  • FIG. 3D illustrates the ER ⁇ degradative activity of exemplary compounds 161a, 31a, and 17a of the present disclosure in an MCF7 cell line 6 hours after administration.
  • FIG. 4A illustrates the ER ⁇ degradative activity of exemplary compounds 54 and 64 of the present disclosure in a CAMA1 cell line 6 hours after administration.
  • FIG. 4B illustrates the ER ⁇ degradative activity of exemplary compounds 54 and 64 of the present disclosure at various concentrations in a CAMA1 cell line 6 hours after administration.
  • FIG. 4C illustrates the ER ⁇ degradative activity of an exemplary compound 64 of the present disclosure at a concentration of 100 nM, as a function of time, in a CAMA1 cell line.
  • FIG. 5A illustrates the ER ⁇ degradative activity of exemplary compounds 54 and 64 of the present disclosure at various concentrations in a ZR-75-1 cell line 6 hours after administration.
  • FIG. 5B illustrates the ER ⁇ degradative activity of exemplary compounds 54 and 64 of the present disclosure at various concentrations in a ZR-75-1 cell line 6 hours after administration.
  • FIG. 5C illustrates the ER ⁇ degradative activity of an exemplary compound 64 of the present disclosure at a concentration of 100 nM, as a function of time, in a ZR-75-1 cell line.
  • FIG. 6 illustrates the ER ⁇ degradative activity of exemplary compounds 160a and 160b of the present disclosure in MCF7, T47D, and CAMA-1 cell lines 6 hours after administration.
  • FIG. 7 illustrates the ER ⁇ degradative activity of exemplary compound 160a of the present disclosure at a concentration of 100 nM, as a function of time, in an MCF7 cell line.
  • FIG. 8 illustrates the ER ⁇ degradative activity of an exemplary compound 160a of the present disclosure at a concentration of 100 nM, in the absence and presence of 1 uM proteasome inhibitor epoxomicin, in a T47D cell line, 6 hours after administration.
  • FIG. 9 illustrates the downregulation of PR resulted from ER ⁇ degradation by exemplary compound 160a of the present disclosure in a T47D cell line 24 hours after administration.
  • FIG. 10 illustrates the ER ⁇ degradative activity of exemplary compound 160a of the present disclosure and several published SERDs at a concentration of 30 nM, in T47D and MCF7 cell lines 6 hours after administration.
  • FIG. 11 illustrates lack of IKZF1, IKZF3 and GSPT1 degradative activity of exemplary compound 160a of the present disclosure, in a RAMOS cell line 24 hours after administration.
  • FIG. 12 illustrates inhibition of the ER ⁇ luciferase reporter by an exemplary compound 160a of the present disclosure in a T47D ERE-Luc reporter cell line 24 hours after administration.
  • FIG. 13 illustrates the dose-dependent ER ⁇ degradative activity of exemplary compound 160a of the present disclosure in MCF7 xenograft tumors after three daily doses.
  • FIG. 14 illustrates the dose-dependent growth inhibitory activity of exemplary compound 160a of the present disclosure in MCF7 xenograft tumors after 21 daily doses.
  • cancer refers to diseases, disorders, and conditions that involve abnormal cell growth with the potential to invade or spread to other parts of the body.
  • exemplary cancers include, but are not limited to, breast cancer, lung cancer, ovarian cancer, endometrial cancer, prostate cancer, and esophageal cancer.
  • Subject refers to an animal, such as a mammal, that has been or will be the object of treatment, observation, or experiment. The methods described herein may be useful for both human therapy and veterinary applications. In one embodiment, the subject is a human.
  • treatment refers to an amelioration of a disease or disorder, or at least one discernible symptom thereof.
  • treatment refers to an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient.
  • treatment or “treating” refers to inhibiting the progression of a disease or disorder, either physically, e.g., stabilization of a discernible symptom, physiologically, e.g., stabilization of a physical parameter, or both.
  • treatment or “treating” refers to delaying the onset of a disease or disorder. For example, treating a cholesterol disorder may comprise decreasing blood cholesterol levels.
  • prevention or “preventing” refers to a reduction of the risk of acquiring a given disease or disorder.
  • a dash (“-”) that is not between two letters or symbols is used to indicate a point of attachment for a substituent.
  • —CN is attached through the carbon atom.
  • C 1 -C 6 alkyl is intended to encompass C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1-6 , C 1-5 , C 1-4 , C 1-3 , C 1-2 , C 2-6 , C 2-5 , C 2-4 , C 2-3 , C 3-6 , C 3-5 , C 3-4 , C 4- 6 , C 4-5 , and C 5-6 alkyl.
  • acyl refers to R—C(O)— groups such as, but not limited to, (alkyl)-C(O)—, (alkenyl)-C(O)—, (alkynyl)-C(O)—, (aryl)-C(O)—, (cycloalkyl)-C(O)—, (heteroaryl)-C(O)—, and (heterocyclyl)-C(O)—, wherein the group is attached to the parent molecular structure through the carbonyl functionality.
  • acyl radical which refers to the total number of chain or ring atoms of the, for example, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, or heteroaryl, portion plus the carbonyl carbon of acyl.
  • a C 4 -acyl has three other ring or chain atoms plus carbonyl.
  • alkenyl refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond, such as a straight or branched group of 2-8 carbon atoms, referred to herein as (C 2 -C 8 )alkenyl.
  • alkenyl groups include, but are not limited to, vinyl, allyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, 2-ethylhexenyl, 2-propyl-2-butenyl, and 4-(2-methyl-3-butene)-pentenyl.
  • alkyl refers to a saturated straight or branched hydrocarbon, such as a straight or branched group of 1-8 carbon atoms, referred to herein as (C 1 -C 8 )alkyl.
  • exemplary alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, butyl, isobutyl, t-butyl, pentyl, t-but
  • alkynyl refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon triple bond, such as a straight or branched group of 2-8 carbon atoms, referred to herein as (C 2 -C 8 )alkynyl.
  • exemplary alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, hexynyl, methylpropynyl, 4-methyl-1-butynyl, 4-propyl-2-pentynyl, and 4-butyl-2-hexynyl.
  • aryl refers to a mono-, bi-, or other multi-carbocyclic, aromatic ring system with 5 to 14 ring atoms.
  • the aryl group can optionally be fused to one or more rings selected from aryls, cycloalkyls, heteroaryls, and heterocyclyls.
  • aryl groups of this present disclosure can be substituted with groups selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide, and thioketone.
  • Exemplary aryl groups include, but are not limited to, phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, and naphthyl, as well as benzo-fused carbocyclic moieties such as 5,6,7,8-tetrahydronaphthyl.
  • Exemplary aryl groups also include, but are not limited to, a monocyclic aromatic ring system, wherein the ring comprises 6 carbon atoms, referred to herein as “C 6 -aryl.”
  • cyano refers to —CN.
  • cycloalkyl refers to a saturated or unsaturated cyclic, bicyclic, or bridged bicyclic hydrocarbon group of 3-16 carbons, or 3-8 carbons, referred to herein as “(C 3 -C 8 )cycloalkyl,” derived from a cycloalkane.
  • exemplary cycloalkyl groups include, but are not limited to, cyclohexanes, cyclohexenes, cyclopentanes, and cyclopentenes.
  • Cycloalkyl groups may be substituted with alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
  • Cycloalkyl groups can be fused to other cycloalkyl (saturated or partially unsaturated), aryl, or heterocyclyl groups, to form a bicycle, tetracycle, etc.
  • cycloalkyl also includes bridged and spiro-fused cyclic structures which may or may not contain heteroatoms.
  • halo or “halogen” as used herein refer to —F, —Cl, —Br, and/or —I.
  • heteroaryl refers to a mono-, bi-, or multi-cyclic, aromatic ring system containing one or more heteroatoms, for example 1-3 heteroatoms, such as nitrogen, oxygen, and sulfur. Heteroaryls can be substituted with one or more substituents including alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
  • Heteroaryls can also be fused to non-aromatic rings.
  • Illustrative examples of heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrimidyl, pyrazyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3)- and (1,2,4)-triazolyl, pyrazinyl, pyrimidilyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, furyl, phenyl, isoxazolyl, and oxazolyl.
  • heteroaryl groups include, but are not limited to, a monocyclic aromatic ring, wherein the ring comprises 2-5 carbon atoms and 1-3 heteroatoms, referred to herein as “(C 2 -C 5 )heteroaryl.”
  • heterocycle refers to a saturated or unsaturated 3- to 18-membered ring containing one, two, three, or four heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.
  • Heterocycles can be aromatic (heteroaryls) or non-aromatic.
  • Heterocycles can be substituted with one or more substituents including alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
  • substituents including alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocycly
  • Heterocycles also include bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from aryls, cycloalkyls, and heterocycles.
  • Exemplary heterocycles include acridinyl, benzimidazolyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, biotinyl, cinnolinyl, dihydrofuryl, dihydroindolyl, dihydropyranyl, dihydrothienyl, dithiazolyl, furyl, homopiperidinyl, imidazolidinyl, imidazolinyl, imidazolyl, indolyl, isoquinolyl, isothiazolidinyl, isothiazolyl, isoxazolidinyl, isoxazolyl, morpholinyl, oxadiazolyl, o
  • hydroxy and “hydroxyl” as used herein refer to —OH.
  • compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.
  • composition refers to a composition comprising at least one compound as disclosed herein formulated together with one or more pharmaceutically acceptable carriers.
  • prodrugs as used herein represents those prodrugs of the compounds of the present disclosure that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the present disclosure.
  • a discussion is provided in Higuchi et al., “Prodrugs as Novel Delivery Systems,” ACS Symposium Series , Vol. 14, and in Roche, E. B., ed. Bioreversible Carriers in Drug Design , American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference.
  • pharmaceutically acceptable salt(s) refers to salts of acidic or basic groups that may be present in compounds used in the present compositions.
  • Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids.
  • the acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, including but not limited to sulfate, citrate, matate, acetate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate (i
  • Compounds included in the present compositions that include an amino moiety may form pharmaceutically acceptable salts with various amino acids, in addition to the acids mentioned above.
  • Compounds included in the present compositions, that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations.
  • Examples of such salts include alkali metal or alkaline earth metal salts and, particularly, calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts.
  • the compounds of the disclosure may contain one or more chiral centers and/or double bonds and, therefore, exist as stereoisomers, such as geometric isomers, enantiomers or diastereomers.
  • stereoisomers when used herein consist of all geometric isomers, enantiomers or diastereomers. These compounds may be designated by the symbols “R” or “S,” depending on the configuration of substituents around the stereogenic carbon atom.
  • Stereoisomers include enantiomers and diastereomers.
  • enantiomers or diastereomers may be designated “( ⁇ )” in nomenclature, but the skilled artisan will recognize that a structure may denote a chiral center implicitly.
  • an enantiomer or stereoisomer may be provided substantially free of the corresponding enantiomer.
  • the compound is a racemic mixture of (S)- and (R)-isomers.
  • provided herein is a mixture of compounds wherein individual compounds of the mixture exist predominately in an (S)- or (R)-isomeric configuration.
  • the compound mixture has an (S)-enantiomeric excess of greater than about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5%, or more.
  • the compound mixture has an (S)-enantiomeric excess of greater than about 55% to about 99.5%, greater than about 60% to about 99.5%, greater than about 65% to about 99.5%, greater than about 70% to about 99.5%, greater than about 75% to about 99.5%, greater than about 80% to about 99.5%, greater than about 85% to about 99.5%, greater than about 90% to about 99.5%, greater than about 95% to about 99.5%, greater than about 96% to about 99.5%, greater than about 97% to about 99.5%, greater than about 98% to greater than about 99.5%, greater than about 99% to about 99.5%, or more.
  • the compound mixture has an (R)-enantiomeric purity of greater than about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5% or more.
  • the compound mixture has an (R)-enantiomeric excess of greater than about 55% to about 99.5%, greater than about 60% to about 99.5%, greater than about 65% to about 99.5%, greater than about 70% to about 99.5%, greater than about 75% to about 99.5%, greater than about 80% to about 99.5%, greater than about 85% to about 99.5%, greater than about 90% to about 99.5%, greater than about 95% to about 99.5%, greater than about 96% to about 99.5%, greater than about 97% to about 99.5%, greater than about 98% to greater than about 99.5%, greater than about 99% to about 99.5% or more.
  • Individual stereoisomers of compounds of the present disclosure can be prepared synthetically from commercially available starting materials that contain asymmetric or stereogenic centers, or by preparation of racemic mixtures followed by resolution methods well known to those of ordinary skill in the art. These methods of resolution are exemplified by: (1) attachment of a mixture of enantiomers to a chiral auxiliary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary; (2) salt formation employing an optically active resolving agent; or (3) direct separation of the mixture of optical enantiomers on chiral chromatographic columns.
  • Stereoisomeric mixtures can also be resolved into their component stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent.
  • Stereoisomers can also be obtained from stereomerically-pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.
  • Geometric isomers can also exist in the compounds of the present disclosure.
  • the present disclosure encompasses the various geometric isomers and mixtures thereof resulting from the arrangement of substituents around a carbon-carbon double bond or arrangement of substituents around a carbocyclic ring.
  • Substituents around a carbon-carbon double bond are designated as being in the “Z” or “E” configuration wherein the terms “Z” and “E” are used in accordance with IUPAC standards.
  • structures depicting double bonds encompass both the E and Z isomers.
  • Substituents around a carbon-carbon double bond alternatively can be referred to as “cis” or “trans,” where “cis” represents substituents on the same side of the double bond and “trans” represents substituents on opposite sides of the double bond.
  • the arrangements of substituents around a carbocyclic ring are designated as “cis” or “trans.”
  • the term “cis” represents substituents on the same side of the plane of the ring and the term “trans” represents substituents on opposite sides of the plane of the ring.
  • Mixtures of compounds wherein the substituents are disposed on both the same and opposite sides of plane of the ring are designated “cis/trans.”
  • structures described herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of hydrogen by deuterium ( 2 H) or tritium ( 3 H), or the replacement of a carbon by a 13 C- or 14 C-carbon atom are within the scope of this disclosure.
  • Such compounds may be useful as, for example, analytical tools, probes in biological assays, or therapeutic agents.
  • provided herein are compounds of Formula (I), or a tautomer, stereoisomer or a mixture of stereoisomers, pharmaceutically acceptable salt, or hydrate thereof:
  • X 1 and X 2 are each independently selected from C(R 3 ) 2 , NR 4 , O, S, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 ;
  • A is selected from:
  • B is selected from 5- to 6-member cycloalkyl, 5- to 6-member aryl, 5- to 6-member heterocycle, and 5- to 6-member heteroaryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • L* is a linker of 1 to 22 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • R 1 and R 2 are each independently selected from H, C 1 -C 6 alkyl, halo, alkoxy, acyloxy, hydroxy, and sulfhydryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • each R 3 is independently selected from H, C 1 -C 6 alkyl, halo, and hydroxy;
  • each R 4 is independently selected from H, C 1 -C 6 alkyl, and acyl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • each R 5 is independently selected from C 1 -C 6 alkyl, halo, cyano, and hydroxy,
  • the compound of Formula (I) may encompass both the E and Z isomers. In some embodiments, the compound of Formula (I) may be a mixture of trans- and -cis olefin.
  • A may be any organic compound
  • A may be any organic compound
  • A may be any organic compound
  • A may be any organic compound
  • A may be any organic compound
  • A may be any organic compound
  • A may be any organic compound
  • A may be any organic compound
  • X 1 and X 2 are each independently selected from C(R 3 ) 2 , NR 4 , O, S, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 ;
  • B is selected from 5- to 6-member cycloalkyl, 5- to 6-member aryl, 5- to 6-member heterocycle, and 5- to 6-member heteroaryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • C is selected from:
  • L* is a linker of 1 to 22 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • R 1 and R 2 are each independently selected from H, C 1 -C 6 alkyl, halo, alkoxy, acyloxy, hydroxy, and sulfhydryl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • each R 3 is independently selected from H, C 1 -C 6 alkyl, halo, and hydroxy;
  • each R 4 is independently selected from H, C 1 -C 6 alkyl, and acyl, each of which is substituted with 0, 1, 2, or 3 R 5 ;
  • each R 5 is independently selected from C 1 -C 6 alkyl, halo, cyano, and hydroxy,
  • C may be
  • C may be
  • C may b
  • C may be
  • C may be
  • B may be a 5-member heterocycle substituted with 0, 1, 2, or 3 R 5 . In some embodiments, B may be a 5-member heterocycle. In some embodiments, B may be a 5-member heterocycle substituted with 1 R 5 . In some embodiments, R 5 may be C 1 alkyl.
  • B may be a 6-member heterocycle substituted with 0, 1, 2, or 3 R 5 . In some embodiments, B may be a 6-member heterocycle. In some embodiments, B may be a 6-member heterocycle substituted with 1 R 5 . In some embodiments, R 5 may be C 1 alkyl.
  • B may be selected from
  • B may be
  • R 1 and R 2 may each be independently selected from H, C 1 -C 3 alkyl, halo, alkoxy, acyloxy, hydroxy, and sulfhydryl, each of which may be substituted with 0, 1, 2, or 3 R 5 .
  • R 1 and R 2 may each be independently selected from H, C 1 alkyl, halo, and hydroxy, each of which may be substituted with 0, 1, 2, or 3 R 5 .
  • R 1 and R 2 may each be independently H or OH.
  • R 1 may be H. In some embodiments, R 1 may be OH. In some embodiments, R 2 may be H. In some embodiments, R 2 may be OH.
  • R 1 may be OH and R 2 may be H. In some embodiments, R 1 may be H and R 2 may be H. In some embodiments, R 1 may be H and R 2 may be OH. In some embodiments, R 1 may be OH and R 2 may be OH.
  • X 1 and X 2 may each be independently selected from C(R 3 ) 2 , NR 4 , O, S, 5 or 6-member cycloalkyl, 5- or 6-member aryl, 5- or 6-member heterocycle, and 5- or 6-member heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • X 1 and X 2 may each be independently selected from CH 2 , NR 4 , O, S, 5 or 6-member cycloalkyl, 5- or 6-member aryl, 5- or 6-member heterocycle, and 5- or 6-member heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • X 1 may be O. In some embodiments, X 1 may be C(R 3 ) 2 . In some embodiments, R 3 may be H or halo. In some embodiments, halo may be fluoro. In some embodiments, R 3 may be H. In some embodiments, X 1 may be NR 4 . In some embodiments, R 4 may be selected from H, C 1 -C 3 alkyl, and acyl, each of which may be substituted with 0, 1, 2, or 3 R 5 . In some embodiments, R 4 may be H. In some embodiments, R 4 may be C 1 -C 3 alkyl substituted with 0, 1, 2, or 3 R 5 .
  • R 4 may be C 1 alkyl substituted with 0, 1, 2, or 3 R 5 . In some embodiments, R 4 may be C 1 alkyl. In some embodiments, R 4 may be acyl substituted with 0, 1, 2, or 3 R 5 .
  • X 1 may be a 5 or 6-member cycloalkyl. In some embodiments, X 1 may be a 5- or 6-member aryl. In some embodiments, X 1 may be a 5- or 6-member heterocycle. In some embodiments, X 1 may be a 5- or 6-member heteroaryl. In some embodiments, X 1 may be a 5 or 6-member cycloalkyl substituted with 0, 1, 2, or 3 R 5 . In some embodiments, X 1 may be a 5- or 6-member aryl substituted with 0, 1, 2, or 3 R 5 . In some embodiments, X 1 may be a 5- or 6-member heterocycle substituted with 0, 1, 2, or 3 R 5 . In some embodiments, X 1 may be a 5- or 6-member heteroaryl substituted with 0, 1, 2, or 3 R 5 .
  • X 1 may be selected from aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, pyrrolyl, pyridinyl, pyrimidinyl, oxiranyl, oxetanyl, tetrahydrofuranyl, furanyl, pyranyl, tetrahydropyranyl, dioxanyl, imidazolyl, pyrazolyl, oxazole, isoxazole, thiazole, isothiazole, triazole, tetrazole, indole, benzimidazole, benzofuran, benzoxazole, benzothiazole, quinoline, isoquinoline, and quinazoline, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • X 1 may be selected from aziridinyl, azetidinyl, pyrrolidinyl, piperidiny
  • X2 may be selected from
  • L* may be linker of 1 to 16 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • L* may be a linker of 1 to 14 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • L* may be a linker of 1 to 12 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • L* may be a linker of 1 to 10 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • L* may be a linker of 1 to 8 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • L* may be a linker of 1 to 6 carbon atoms in length, wherein one or more carbon atoms are each optionally and independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • L* may be a linker wherein two carbon atoms are each independently replaced by a heterocycle, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • L* may be a linker wherein one carbon atom is replaced by a heterocycle and one carbon atom is replaced by a cycloalkyl, each of which is independently substituted with 0, 1, 2, or 3 R 5 .
  • L* may be a linker wherein more than one carbon atoms are each independently replaced by a group selected from C(O), O, NR 4 , S, C 2 -alkenyl, C 2 -alkynyl, cycloalkyl, aryl, heterocycle, and heteroaryl, each of which is substituted with 0, 1, 2, or 3 R 5 .
  • L* may be a linker wherein more than one carbon atoms are each independently replaced by a group selected from C(O), 0, and NR 4 , each of which is substituted with 0, 1, 2, or 3 R 5 .
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • L* may be
  • provided herein is a compound, or tautomer, stereoisomer, pharmaceutically acceptable salt, or hydrate thereof, chosen from:
  • provided herein is a compound, or tautomer, stereoisomer, pharmaceutically acceptable salt, or hydrate thereof, chosen from:
  • provided herein is a compound, or tautomer, stereoisomer, pharmaceutically acceptable salt, or hydrate thereof, chosen from:
  • provided herein is a compound, or tautomer, stereoisomer, pharmaceutically acceptable salt, or hydrate thereof, chosen from:
  • provided herein is a compound, or tautomer, stereoisomer, pharmaceutically acceptable salt, or hydrate thereof, chosen from:
  • provided herein is a compound, or tautomer, stereoisomer, pharmaceutically acceptable salt, or hydrate thereof, chosen from:
  • provided herein is a compound, or tautomer, stereoisomer, or pharmaceutically acceptable salt, or hydrate thereof, chosen from:
  • provided herein is a compound, or tautomer, stereoisomer, or pharmaceutically acceptable salt, or hydrate thereof, chosen from:
  • provided herein is a pharmaceutically acceptable salt of a compound of Formula (I), or (II). In some embodiments, provided herein is a compound of Formula (I), or (II).
  • provided herein is a compound, or pharmaceutically acceptable salt thereof, chosen from the compounds listed in Table 1.
  • compositions of the present disclosure comprise at least one compound of Formulae (I) or (II), or tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof formulated together with one or more pharmaceutically acceptable carriers.
  • These formulations include those suitable for oral, rectal, topical, buccal and parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous) administration. The most suitable form of administration in any given case will depend on the degree and severity of the condition being treated and on the nature of the particular compound being used.
  • Formulations suitable for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of a compound of the present disclosure as powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil emulsion.
  • such formulations may be prepared by any suitable method of pharmacy which includes the step of bringing into association at least one compound of the present disclosure as the active compound and a carrier or excipient (which may constitute one or more accessory ingredients).
  • the carrier must be acceptable in the sense of being compatible with the other ingredients of the formulation and must not be deleterious to the recipient.
  • the carrier may be a solid or a liquid, or both, and may be formulated with at least one compound described herein as the active compound in a unit-dose formulation, for example, a tablet, which may contain from about 0.05% to about 95% by weight of the at least one active compound.
  • a unit-dose formulation for example, a tablet, which may contain from about 0.05% to about 95% by weight of the at least one active compound.
  • Other pharmacologically active substances may also be present including other compounds.
  • the formulations of the present disclosure may be prepared by any of the well-known techniques of pharmacy consisting essentially of admixing the components.
  • liquid compositions conventional nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • Liquid pharmacologically administrable compositions can, for example, be prepared by, for example, dissolving or dispersing, at least one active compound of the present disclosure as described herein and optional pharmaceutical adjuvants in an excipient, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form a solution or suspension.
  • suitable formulations may be prepared by uniformly and intimately admixing the at least one active compound of the present disclosure with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • a tablet may be prepared by compressing or molding a powder or granules of at least one compound of the present disclosure, which may be optionally combined with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing, in a suitable machine, at least one compound of the present disclosure in a free-flowing form, such as a powder or granules, which may be optionally mixed with a binder, lubricant, inert diluent and/or surface active/dispersing agent(s).
  • Molded tablets may be made by molding, in a suitable machine, where the powdered form of at least one compound of the present disclosure is moistened with an inert liquid diluent.
  • Formulations suitable for buccal (sub-lingual) administration include lozenges comprising at least one compound of the present disclosure in a flavored base, usually sucrose and acacia or tragacanth, and pastilles comprising the at least one compound in an inert base such as gelatin and glycerin or sucrose and acacia.
  • Formulations of the present disclosure suitable for parenteral administration comprise sterile aqueous preparations of at least one compound of Formula (I) or (II), or tautomers, stereoisomers, pharmaceutically acceptable salts, and hydrates thereof, which are approximately isotonic with the blood of the intended recipient.
  • These preparations are administered intravenously, although administration may also be effected by means of subcutaneous, intramuscular, or intradermal injection.
  • Such preparations may conveniently be prepared by admixing at least one compound described herein with water and rendering the resulting solution sterile and isotonic with the blood.
  • Injectable compositions according to the present disclosure may contain from about 0.1 to about 5% w/w of the active compound.
  • Formulations suitable for rectal administration are presented as unit-dose suppositories. These may be prepared by admixing at least one compound as described herein with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
  • Formulations suitable for topical application to the skin may take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil.
  • Carriers and excipients which may be used include Vaseline, lanoline, polyethylene glycols, alcohols, and combinations of two or more thereof.
  • the active compound i.e., at least one compound of Formula (I) or (II), or tautomers, stereoisomers, pharmaceutically acceptable salts, and hydrates thereof
  • the active compound i.e., at least one compound of Formula (I) or (II), or tautomers, stereoisomers, pharmaceutically acceptable salts, and hydrates thereof
  • the amount of active compound administered may be dependent on the subject being treated, the subject's weight, the manner of administration and the judgment of the prescribing physician.
  • a dosing schedule may involve the daily or semi-daily administration of the encapsulated compound at a perceived dosage of about 1 ⁇ g to about 1000 mg.
  • intermittent administration such as on a monthly or yearly basis, of a dose of the encapsulated compound may be employed.
  • Encapsulation facilitates access to the site of action and allows the administration of the active ingredients simultaneously, in theory producing a synergistic effect.
  • physicians will readily determine optimum dosages and will be able to readily modify administration to achieve such dosages.
  • a therapeutically effective amount of a compound or composition disclosed herein can be measured by the therapeutic effectiveness of the compound.
  • the dosages may be varied depending upon the requirements of the patient, the severity of the condition being treated, and the compound being used.
  • the therapeutically effective amount of a disclosed compound is sufficient to establish a maximal plasma concentration.
  • Preliminary doses as, for example, determined according to animal tests, and the scaling of dosages for human administration is performed according to art-accepted practices.
  • Toxicity and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED 50 .
  • Compositions that exhibit large therapeutic indices are preferable.
  • Therapeutically effective dosages achieved in one animal model may be converted for use in another animal, including humans, using conversion factors known in the art (see, e.g., Freireich et al., Cancer Chemother. Reports 50(4):219-244 (1966) and the following Table for Equivalent Surface Area Dosage Factors).
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • a therapeutically effective amount may vary with the subject's age, condition, and gender, as well as the severity of the medical condition in the subject.
  • the dosage may be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof is administered to treat cancer in a subject in need thereof.
  • the cancer is chosen from breast cancer, lung cancer, ovarian cancer, endometrial cancer, prostate cancer, and esophageal cancer.
  • the cancer is breast cancer.
  • the cancer is lung cancer.
  • the cancer is ovarian cancer.
  • the cancer is endometrial cancer.
  • the cancer is prostate cancer.
  • the cancer is esophageal cancer.
  • the cancer is positive for ER ⁇ .
  • a compound of Formulae (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof is administered as a pharmaceutical composition.
  • the subject has been previously treated with tamoxifen.
  • the therapeutic treatment is for the treatment of breast cancer, lung cancer, ovarian cancer, endometrial cancer, prostate cancer, and esophageal cancer.
  • the therapeutic treatment is for the treatment of breast cancer.
  • the therapeutic treatment is for lung cancer.
  • the therapeutic treatment is for the treatment of ovarian cancer.
  • the therapeutic treatment is for the treatment of endometrial cancer.
  • the therapeutic treatment is for the treatment of prostate cancer.
  • the therapeutic treatment is for the treatment of esophageal cancer. In some embodiments, the therapeutic treatment is for the treatment of estrogen-related diseases and conditions. In some embodiments, the therapeutic treatment is for the treatment of infertility. In some embodiments, the therapeutic treatment is for the treatment of ovulatory dysfunction. In some embodiments, the therapeutic treatment is for the treatment of postmenopausal osteoporosis. In some embodiments, the therapeutic treatment is for the treatment of estrogen-related gynecomastia. In some embodiments, the therapeutic treatment is for the treatment of dyspareunia due to menopause. In some embodiments, the therapeutic treatment is for the treatment of retroperitoneal fibrosis. In some embodiments, the therapeutic treatment is for the treatment of idiopathic sclerosing mesenteritis.
  • provided herein is a use of a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof, in the preparation of a medicament.
  • a method of inhibiting cell growth comprising contacting a cell with a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof.
  • the cell may express ER ⁇ .
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof is administered in combination with another therapeutic agent.
  • the other therapeutic agent can provide additive or synergistic value relative to the administration of a compound of the present disclosure alone.
  • the therapeutic agent can be selected from, for example, hormones and hormonal analogues; signal transduction pathway inhibitors; topoisomerase I inhibitors; topoisomerase II inhibitors; antimetabolite neoplastic agents; antibiotic neoplastic agents; alkylating agents; anti-microtubule agents; platinum coordination complexes; aromatase inhibitors; and anti-mitotic agents.
  • the therapeutic agent may be a hormone or hormonal analogue. In some embodiments, the therapeutic agent may be a signal transduction pathway inhibitor. In some embodiments, the therapeutic agent may be a topoisomerase I inhibitor. In some embodiments, the therapeutic agent may be a topoisomerase II inhibitor. In some embodiments, the therapeutic agent may be an antimetabolite neoplastic agent. In some embodiments, the therapeutic agent may be an antibiotic neoplastic agent. In some embodiments, the therapeutic agent may be an alkylating agent. In some embodiments, the therapeutic agent may be an anti-microtubule agent. In some embodiments, the therapeutic agent may be a platinum coordination complex. In some embodiments, the therapeutic agent may be an aromatase inhibitor. In some embodiments, the therapeutic agent may be an anti-mitotic agent.
  • the aromatase inhibitor may be selected from anastrazole, letrozole, vorozole, fadrozole, exemestane, and formestane.
  • the aromatase inhibitor is anastrazole.
  • the aromatase inhibitor may be letrozole.
  • the aromatase inhibitor may be vorozole.
  • the aromatase inhibitor may be fadrozole.
  • the aromatase inhibitor may be exemestane.
  • the aromatase inhibitor may be formestane.
  • the anti-mitotic agent may be selected from paclitaxel, docetaxel, and Abraxane. In some embodiments, the anti-mitotic agent may be paclitaxel. In some embodiments, the anti-mitotic agent may be docetaxel. In some embodiments, the anti-mitotic agent may be Abraxane.
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof may be administered in combination with a hormone or hormonal analog.
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof may be administered in combination with a signal transduction pathway inhibitor.
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof may be administered in combination with an antimetabolite neoplastic agent.
  • a compound of Formulae (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof may be administered in combination with a topoisomerase I inhibitor.
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof may be administered in combination with a topoisomerase II inhibitor.
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof may be administered in combination with an aromatase inhibitor.
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof may be administered in combination with one or more anti-cancer agents.
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof may be administered in combination with an anti-cancer agent, wherein the anti-cancer agent is tamoxifen.
  • a compound of Formula (I) or (II), or a tautomer, stereoisomer, pharmaceutically acceptable salt or hydrate thereof may be administered in combination with an anti-cancer agent, wherein the anti-cancer agent is fulvestrant.
  • the chemical entities described herein can be synthesized according to one or more illustrative schemes herein and/or techniques well known in the art. Unless specified to the contrary, the reactions described herein take place at atmospheric pressure, generally within a temperature range from about ⁇ 10° C. to about 200° C. Further, except as otherwise specified, reaction times and conditions are intended to be approximate, e.g., taking place at about atmospheric pressure within a temperature range of about ⁇ 10° C. to about 200° C. over a period that can be, for example, about 1 to about 24 hours; reactions left to run overnight in some embodiments can average a period of about 16 hours.
  • Isolation and purification of the chemical entities and intermediates described herein can be effected, if desired, by any suitable separation or purification procedure such as, for example, filtration, extraction, crystallization, column chromatography, thin-layer chromatography or thick-layer chromatography, or a combination of these procedures.
  • any suitable separation or purification procedure such as, for example, filtration, extraction, crystallization, column chromatography, thin-layer chromatography or thick-layer chromatography, or a combination of these procedures.
  • suitable separation or purification procedure such as, for example, filtration, extraction, crystallization, column chromatography, thin-layer chromatography or thick-layer chromatography, or a combination of these procedures.
  • protecting groups for sensitive or reactive groups may be employed where necessary, in accordance with general principles of chemistry.
  • Protecting groups are manipulated according to standard methods of organic synthesis (T.W. Greene and P.G.M. Wuts (1999) Protective Groups in Organic Synthesis, 3 rd Ed., John Wiley & Sons). These groups may be removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art.
  • the (R)- and (S)-isomers of the nonlimiting exemplary compounds can be resolved by methods known to those skilled in the art, for example, by formation of diastereoisomeric salts or complexes which can be separated, e.g., by crystallization; via formation of diastereoisomeric derivatives which can be separated, e.g., by crystallization, gas-liquid or liquid chromatography; selective reaction of one enantiomer with an enantiomer-specific reagent, e.g., enzymatic oxidation or reduction, followed by separation of the modified and unmodified enantiomers; or gas-liquid or liquid chromatography in a chiral environment, e.g., on a chiral support, such as silica with a bound chiral ligand or in the presence of a chiral solvent.
  • a specific enantiomer can be synthesized by asymmetric synthesis using optically active reagents
  • the compounds described herein can be optionally contacted with a pharmaceutically acceptable acid to form the corresponding acid addition salts. Also, the compounds described herein can be optionally contacted with a pharmaceutically acceptable base to form the corresponding basic addition salts.
  • disclosed compounds can generally be synthesized by an appropriate combination of generally well-known synthetic methods. Techniques useful in synthesizing these chemical entities are both readily apparent and accessible to those of skill in the relevant art, based on the instant disclosure. Many of the optionally substituted starting compounds and other reactants are commercially available, e.g., from Millipore Sigma or can be readily prepared by those skilled in the art using commonly employed synthetic methodology.
  • HPLC spectra for all compounds were acquired using an Agilent 1200 Series system with DAD detector. Chromatography was performed on a 2.1 ⁇ 150 mm Zorbax 300SB-C18 5 ⁇ m column with water containing 0.1% formic acid as solvent A and acetonitrile containing 0.1% formic acid as solvent B at a flow rate of 0.4 mL/min. The gradient program was as follows: 1% B (0-1 min), 1-99% B (1-4 min), and 99% B (4-8 min). High-resolution mass spectra (HRMS) data were acquired in positive ion mode using an Agilent G1969A API-TOF with an electrospray ionization (ESI) source.
  • HRMS high-resolution mass spectra
  • Step 4 Synthesis of tert-butyl 4-(2-(4-(2-phenyl-1-(4-(pivaloyloxy) phenyl)but-1-enyl)phenoxy)ethyl)piperazine-1-carboxylate
  • LCMS indicated a ratio to 2/3, M/Z 613.5 (M+1)+.
  • Step 5 Synthesis of 4-(2-phenyl-1-(4-(2-(piperazin-1-yl)ethoxy)phenyl)but-1-enyl)phenol Hydrochloride (Intermediate 1)
  • Step 1 Synthesis of 4-(2-phenyl-1-(4-(((trifluoromethyl)sulfonyl)oxy)phenyl)but-1-en-1-yl)phenyl Pivalate
  • Step 2 Synthesis of tert-butyl ((1-(4-(1-(4-hydroxyphenyl)-2-phenylbut-1-en-1-yl)phenyl)piperidin-4-yl)methyl)carbamate
  • Step 3 Synthesis of 4-(1-(4-(4-(aminomethyl)piperidin-1-yl)phenyl)-2-phenylbut-1-en-1-yl)phenol Hydrochloride (Intermediate 2)
  • Step 3 Synthesis of 4-(1-(4-(2-(methylamino)ethoxy)phenyl)-2-phenylbut-1-en-1-yl)phenol (Intermediate 3) and 4-(1-(4-(2-aminoethoxy)phenyl)-2-phenylbut-1-en-1-yl)phenol (Intermediate 4)
  • Step 2 Synthesis of 4-(2-phenyl-1-(4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)but-1-en-1-yl)phenol
  • Step 3 Synthesis of 2-(4-(1-(4-((5,5-dimethoxypentyl)oxy)phenyl)-2-phenylbut-1-en-1-yl)phenoxy)tetrahydro-2H-pyran
  • Step 1 Synthesis of 4-(2-phenyl-1-(4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)but-1-en-1-yl)phenyl trifluoromethanesulfonate
  • Step 2a Synthesis of 4-(dimethoxymethyl)-1-(4-(2-phenyl-1-(4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)but-1-en-1-yl)phenyl)piperidine (Intermediate 6)
  • Step 2b Synthesis of 4-(2,2-dimethoxyethyl)-1-(4-(2-phenyl-1-(4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)but-1-en-1-yl)phenyl)piperidine (Intermediate 7)
  • This compound was synthesized using the same method as described in Step 2a except 4-(2,2-dimethoxyethyl)piperidine was used.
  • the desired 4-(2,2-dimethoxyethyl)-1-(4-(2-phenyl-1-(4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)but-1-en-1-yl)phenyl)piperidine was obtained as a gummy material (20.8% yield, 94.5% purity).
  • Step 1 Preparation of 1-(4-(1-(4-hydroxyphenyl)-2-phenylbut-1-en-1-yl)phenyl)piperidine-4-carbaldehyde
  • Step 2 Preparation of (S)-3-(5-(4-((1-(4-(1-(4-(1-(4-hydroxyphenyl)-2-phenylbut-1-en-1-yl)phenyl)piperidin-4-yl)methyl)piperazin-1-yl)-1-oxoisoindolin-2-yl)piperidine-2,6-dione (160a)
  • Step 1 through Step 6 Synthesis of (E)-1-(4-(1-(4-hydroxyphenyl)-2-phenylbut-1-en-1-yl)phenyl)piperidine-4-carbaldehyde (160b-int-6)
  • 160b-int-6 1H-NMR (CDCl3, 400 MHz) ⁇ 9.59 (s, 1H, aldehyde), 7.01-7.11 (m, 7H), 6.72 (d, 2H), 6.65 (d, 2H), 6.49 (d, 2H), 4.92 (br s, 1H, phenol), 3.43 (m, 2H), 2.67 (m, 2H), 2.36 (q, 2H), 2.25 (m, 1H), 1.88 (m, 2H), 1.63 (m, 2H), 0.84 (t, 3H).
  • Step 7 Synthesis of (S,E)-3-(5-(4-((1-(4-(1-(4-(1-(4-hydroxyphenyl)-2-phenylbut-1-en-1-yl)phenyl)piperidin-4-yl)methyl)piperazin-1-yl)-1-oxoisoindolin-2-yl)piperidine-2,6-dione (160b)
  • Step 1 Synthesis of 2-(4-(1-(4-(3-(1,3-dioxolan-2-yl)propoxy)phenyl)-2-phenylbut-1-en-1-yl)phenoxy)tetrahydro-2H-pyran
  • Step 2 Synthesis of 4-(4-(1-(4-hydroxyphenyl)-2-phenylbut-1-en-1-yl)phenoxy)butanal
  • Step 3 Synthesis of (S)-3-(5-(4-(4-(4-(4-(1-(4-hydroxyphenyl)-2-phenylbut-1-en-1-yl)phenoxy)butyl)piperazin-1-yl)-1-oxoisoindolin-2-yl)piperidine-2,6-dione (17a)
  • N-Desmethyltamoxifen (0.1 mmol) was dissolved in DMSO/DCM (1 mL/5 mL) followed by addition of NMM (0.2 mmol), Linker (0.1 mmol), HOAt (0.15 mmol) and EDCl (0.15 mmol). The mixture was allowed to stir at room temperature overnight. The progress of the reaction was monitored by LCMS. The crude product was then purified by prep-HPLC to yield the desired product.
  • T47D cells were plated in 24-well plates at 1.5 ⁇ 10 5 cells/well in the RPMI growth medium containing 10% FBS and 1 ⁇ Penicillin Streptomycin, and then incubated at 37° C. overnight. The following day, the test compound was administered to the cells by using 1000 ⁇ compound stock solution prepared in DMSO at various concentrations. After administration of the compound, the cells were then incubated at 37 C for various period of treatment time, e.g., as indicated in the figures.
  • the membrane was then incubated with primary antibodies (mouse anti-ER ⁇ (1:500, Santa Cruz), mouse anti-GAPDH (1:5,000, Santa Cruz), or mouse anti-actin (1:4,000, Santa Cruz)) overnight at 4° C., followed by washing (3 times) with TBS-T, and then incubation with horseradish peroxidase-conjugated rabbit anti-mouse IgG (1:5,000) for 60 minutes. After the TBS-T washes, blots were developed with an enhanced chemiluminescence kit (ThermoFisher Scientific) and the bands were imaged and quantified by densitometry (Bio-Rad).
  • primary antibodies mouse anti-ER ⁇ (1:500, Santa Cruz
  • mouse anti-GAPDH (1:5,000, Santa Cruz
  • mouse anti-actin 1:4,000, Santa Cruz
  • FIGS. 2A, 2B, 2C, and 2D show the ER ⁇ degradation ability of compounds 8, 15, 54, 64, 88, 89, 108, 120, 126, and 143 in T47D cells at various concentrations and time points.
  • FIG. 2E shows the ER ⁇ degradation ability of compound 64 in T47D cells at a concentration of 100 nM at various time points.
  • the DC 50 values (i.e., the concentration of test compound at which 50% of the target protein is degraded) was calculated for various compounds of the present disclosure and are shown in the Table 3, below.
  • A corresponds to a DC 50 value less than or equal to 1.5 ⁇ M;
  • B corresponds to a DC 50 value greater than 1.5 ⁇ M and less than 3 ⁇ M; and
  • C corresponds to a DC 50 value greater than 3 ⁇ M.
  • MCF7 cells were plated in 24-well plates at 1.5 ⁇ 10 5 cells/well in the DMEM growth medium containing 10% FBS and 1 ⁇ Penicillin Streptomycin, and then incubated at 37° C. overnight. The following day, the test compound was administered to the cells by using 1000 ⁇ compound stock solution prepared in DMSO at various concentrations. After administration of the compound, the cells were then incubated at 37° C. for various period of treatment time, e.g., as indicated in the figures.
  • FIGS. 3A and 3B show the ER ⁇ degradation ability of compounds 15, 54, and 64 in MCF7 cells at various concentrations at 4 and 6 hours, respectively.
  • FIG. 3C shows the ER ⁇ degradation ability of exemplary compounds 160a, 184a, 17a, 16a, 31a, 28a, 32a, 29a, 161a, and 185a of the present disclosure in an MCF7 cell line 6 hours after administration.
  • FIG. 3D shows the ER ⁇ degradation ability of exemplary compounds 161a, 31a, and 17a of the present disclosure in an MCF7 cell line 6 hours after administration.
  • CAMA1 cells were plated in 24-well plates at 2 ⁇ 10 5 cells/well in the RPMI growth medium containing 20% FBS and 1 ⁇ Penicillin Streptomycin, and then incubated at 37° C. overnight. The following day, the test compound was administered to the cells by using 1000 ⁇ compound stock solution prepared in DMSO at various concentrations. After administration of the compound, the cells were then incubated at 37° C. for various period of treatment time, e.g., as indicated in the figures.
  • the membrane was then incubated with primary antibodies (mouse anti-ER ⁇ (1:500, Santa Cruz), mouse anti-GAPDH (1:5,000, Santa Cruz), or mouse anti-actin (1:4,000, Santa Cruz)) overnight at 4° C., followed by washing (3 times) with TBS-T, and then incubation with horseradish peroxidase-conjugated rabbit anti-mouse IgG (1:5,000) for 60 minutes. After the TBS-T washes, blots were developed with an enhanced chemiluminescence kit (ThermoFisher Scientific) and the bands were imaged and quantified by densitometry (Bio-Rad).
  • primary antibodies mouse anti-ER ⁇ (1:500, Santa Cruz
  • mouse anti-GAPDH (1:5,000, Santa Cruz
  • mouse anti-actin 1:4,000, Santa Cruz
  • FIGS. 4A and 4B show the ER ⁇ degradation ability of compounds 54 and 64 in CAMA1 cells at various concentrations at 6 hours after administration.
  • FIG. 4C shows the ER ⁇ degradation ability of compound 64 at a concentration of 100 nM at various time points in CAMA1 cells.
  • ZR-75-1 cells were plated in 24-well plates at 1.8 ⁇ 10 5 cells/well in the RPMI growth medium containing 20% FBS and 1 ⁇ Penicillin Streptomycin, and then incubated at 37° C. overnight. The following day, the test compound was administered to the cells by using 1000 ⁇ compound stock solution prepared in DMSO at various concentrations. After administration of the compound, the cells were then incubated at 37° C. for various period of treatment time, e.g., as indicated in the figures.
  • the membrane was then incubated with primary antibodies (mouse anti-ER ⁇ (1:500, Santa Cruz), mouse anti-GAPDH (1:5,000, Santa Cruz), or mouse anti-actin (1:4,000, Santa Cruz)) overnight at 4° C., followed by washing (3 times) with TBS-T, and then incubation with horseradish peroxidase-conjugated rabbit anti-mouse IgG (1:5,000) for 60 minutes. After the TBS-T washes, blots were developed with an enhanced chemiluminescence kit (ThermoFisher Scientific) and the bands were imaged and quantified by densitometry (Bio-Rad).
  • primary antibodies mouse anti-ER ⁇ (1:500, Santa Cruz
  • mouse anti-GAPDH (1:5,000, Santa Cruz
  • mouse anti-actin 1:4,000, Santa Cruz
  • FIGS. 5A and 5B show the ER ⁇ degradation ability of compounds 54 and 64 in ZR-75-1 cells at various concentrations at 6 hours after administration.
  • FIG. 5 C shows the ER ⁇ degradation ability of compound 64 at a concentration of 100 nM at various time points in ZR-75-1 cells.
  • Proteins in cell lysate were separated by SDS-PAGE and transferred to Odyssey nitrocellulose membranes (Licor) with Iblot® dry blotting transfer system (ThermoFisher). Nonspecific binding was blocked by incubating membranes with Intercept Blocking Buffer (Licor) for 1 hour at room temperature with gentle shaking. The membranes were then incubated overnight at 4° C.
  • FIG. 6 shows the ER ⁇ degradation ability of compounds 160a and 160b in MCF7, T47D, and CAMA-1 cell lines at various concentrations 6 hours after administration.
  • FIG. 7 shows the ER ⁇ degradation ability of exemplary compound 160a of the present disclosure at a concentration of 100 nM, as a function of time, in a MCF7 cell line.
  • FIG. 8 shows the ER ⁇ degradation ability of exemplary compound 160a of the present disclosure at a concentration of 100 nM, in the absence and presence of 1 uM proteasome inhibitor epoxomicin, in a T47D cell line, 6 hours after administration.
  • FIG. 9 shows the downregulation of PR resulted from ER ⁇ degradation by exemplary compound 160a of the present disclosure in a T47D cell line 24 hours after administration.
  • FIG. 10 shows the ER ⁇ degradation ability of exemplary compound 160a of the present disclosure and several published SERDs at a concentration of 30 nM, in T47D and MCF7 cell lines 6 hours after administration.
  • FIG. 11 shows the lack of IKZF1, IKZF3 and GSPT1 degradation ability of exemplary compound 160a of the present disclosure, in a RAMOS cell line 24 hours after administration. Lenalidomide and CC-885 were used as controls.
  • Example 7 Growth Inhibitory Activity of Exemplary Compound 160a of the Present Disclosure in MCF7, T47D, CAMA-1, and RAMOS
  • MCF7 (ATCC), T47D(ATCC) and CAMA-1 (ATCC) were plated in 96-well plates at 3,000 cell/well in 90 ul of RPMI growth medium containing 10% FBS and 1% Penicillin Streptomycin.
  • RAMOS (ATCC) cells were plated in 96-well plates at 5,000 cell/well in 90 ul of RPMI growth medium containing 10% heat inactive serum and 1% Penicillin Streptomycin. Cells were incubated at 37° C. overnight. The following day, the test compound was administered to the cells by using 10 ⁇ compound stock solution prepared in growth medium at various concentrations. After administration of the compound, the MCF7, T47D, CAMA-1 cells were then incubated at 37° C. for 6 days.
  • RAMOS cells were incubated for 3 days. Before CellTiter-Glo assay, the plates were equilibrated at room temperature for approximately 10 minutes. 100 ul of CellTiter-Glo® Reagent (Promega) was added to each well. The plates were then incubated at room temperature for 10 minutes and luminescence was recorded by EnSpire plate reader (PerkinElmer).
  • Table 4 shows the growth inhibitory activity (see EC50 value) of exemplary compound 160a of the present disclosure in 4 cell lines (i.e. MCF7, T47D, CAMA-1, and RAMOS) 6 days after administration.
  • Example 8 Inhibition of the ER ⁇ Luciferase Reporter by Compound 160a in a T47D ERE-Luc Reporter Cell Line 24 Hours after Administration
  • T47D-KBluc (ATCC) cells were plated in 96-well plates at 20,000 cells/well in 90 ul of RPMI growth medium containing 10% FBS, 5 ⁇ g/ml bovine insulin and 1% Penicillin Streptomycin, and then incubated at 37° C. overnight. The following day, the test compound was administered to the cells by using 10 ⁇ compound stock solution prepared in growth medium at various concentrations. After administration of the compound, the cells were then incubated at 37° C. for 24 hours. Before One-Glo assay, the plate was equilibrated at room temperature for approximately 10 minutes. 100 ul of One-Glo® Reagent (Promega) was added to each well. The plates were then incubated at room temperature for 10 minutes and luminescence was recorded by EnSpire plate reader (PerkinElmer).
  • FIG. 12 shows inhibition of the ER ⁇ luciferase reporter by an exemplary compound 160a of the present disclosure in a T47D ERE-Luc reporter cell line 24 hours after administration.
  • MCF-7 tumor bearing BALB/c nude mice were randomized when the average tumor volume reaches approximately 300-500 mm3, with 5 mice per treatment group. Dosing regimen for each group was indicated in FIG. 13 . 8 hours after the last dose, tumors were collected and snap frozen in liquid nitrogen. Tumor lysates were prepared by grinding tumors in RIPA Buffer (contains 1% Protease Inhibitor Cocktail and 1% Phosphatase Inhibitor Cocktail 2) with Tissuelyser LT at 50 Hz for 5 min. Proteins in tumor lysates were separated by SDS-PAGE and transferred to Odyssey nitrocellulose membranes with Iblot® dry blotting transfer system (ThermoFisher).
  • Nonspecific binding was blocked by incubation with blocking buffer (5% milk in TBS-T) at room temperature for 60 min.
  • the membrane was then incubated with primary antibodies, anti-ER ⁇ and anti-GAPDH, overnight at 4 C. The following day, the membranes were washed 5 times with TBS-T, and then incubated with horseradish peroxidase-conjugated secondary antibody for 60 min. After TBS-T washes, blots were developed with West Femto Maximum Sensitivity kit (thermo fisher scientific). The bands were imaged and quantified by Amersham Imager 680.
  • FIG. 13 shows the dose-dependent ER ⁇ degradation ability of exemplary compound 160a of the present disclosure in MCF7 xenograft tumors after three daily doses. Tumors were collected 8 hours after last dose. Fulvestrant at indicated dose was used as comparison.
  • Example 10 Inhibitory Activity of Exemplary Compound 160a of the Present Disclosure in MCF7 Xenograft Tumors
  • MCF-7 tumor bearing mice BALB/c nude mice were randomized when the average tumor volume reaches approximately 150-200 mm 3 , with 10 mice per treatment group. Dosing regimen for each group is indicated in FIG. 14 . Tumor weight was measured twice every week.
  • FIG. 14 shows the dose-dependent growth inhibitory activity of exemplary compound 160a of the present disclosure in MCF7 xenograft tumors after 21 daily doses. Fulvestrant at indicated dose was used as comparison.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Indole Compounds (AREA)
US16/690,611 2018-11-21 2019-11-21 Compounds having estrogen receptor alpha degradation activity and uses thereof Active US10696659B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/690,611 US10696659B2 (en) 2018-11-21 2019-11-21 Compounds having estrogen receptor alpha degradation activity and uses thereof
US16/851,994 US11117885B2 (en) 2018-11-21 2020-04-17 Compounds having estrogen receptor alpha degradation activity and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862770476P 2018-11-21 2018-11-21
US16/690,611 US10696659B2 (en) 2018-11-21 2019-11-21 Compounds having estrogen receptor alpha degradation activity and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/851,994 Continuation US11117885B2 (en) 2018-11-21 2020-04-17 Compounds having estrogen receptor alpha degradation activity and uses thereof

Publications (2)

Publication Number Publication Date
US20200157078A1 US20200157078A1 (en) 2020-05-21
US10696659B2 true US10696659B2 (en) 2020-06-30

Family

ID=68653428

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/690,611 Active US10696659B2 (en) 2018-11-21 2019-11-21 Compounds having estrogen receptor alpha degradation activity and uses thereof
US16/851,994 Active US11117885B2 (en) 2018-11-21 2020-04-17 Compounds having estrogen receptor alpha degradation activity and uses thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/851,994 Active US11117885B2 (en) 2018-11-21 2020-04-17 Compounds having estrogen receptor alpha degradation activity and uses thereof

Country Status (7)

Country Link
US (2) US10696659B2 (fr)
EP (1) EP3675839B1 (fr)
CN (2) CN113164409B (fr)
AU (1) AU2019384807A1 (fr)
CA (1) CA3120530A1 (fr)
SG (1) SG11202105256QA (fr)
WO (1) WO2020106933A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11642342B2 (en) 2019-12-23 2023-05-09 Accutar Biotechnology Combinations of estrogen receptor degraders and cyclin-dependent kinase inhibitors for treating cancer

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114085212B (zh) * 2018-09-30 2023-05-02 中国科学院上海药物研究所 异吲哚啉类化合物、其制备方法、药物组合物及用途
US11351149B2 (en) 2020-09-03 2022-06-07 Pfizer Inc. Nitrile-containing antiviral compounds
US20240058318A1 (en) * 2020-12-10 2024-02-22 Mayo Foundation For Medical Education And Research Degrading pkcb1 to treat cancer
WO2022217010A1 (fr) * 2021-04-09 2022-10-13 Endotarget Inc. Composés et procédés pour la dégradation ciblée de récepteurs d'œstrogène
KR102503296B1 (ko) 2021-05-18 2023-02-24 재단법인 대구경북첨단의료산업진흥재단 신규 에텐 화합물 및 이를 유효성분으로 포함하는 암의 예방 또는 치료용 약학적 조성물
WO2022245124A1 (fr) 2021-05-18 2022-11-24 재단법인 대구경북첨단의료산업진흥재단 Nouveau composé d'éthène et composition pharmaceutique le comprenant en tant que principe actif pour la prévention ou le traitement du cancer
EP4367112A1 (fr) 2021-07-09 2024-05-15 Plexium, Inc. Composés aryle et compositions pharmaceutiques modulant l'ikzf2
EP4378936A1 (fr) * 2021-07-30 2024-06-05 Hinova Pharmaceuticals Inc. Composé hétérocyclique chimère bifonctionnel et son utilisation en tant qu'agent de dégradation du récepteur des androgènes
EP4377318A1 (fr) * 2021-07-30 2024-06-05 BeiGene, Ltd. Composés bifonctionnels à base de pyrrolo [2, 3-b] pyrazine utilisés comme agents de dégradation de la hpk1 et leur utilisation
CN115894450B (zh) * 2021-11-30 2023-09-12 山东如至生物医药科技有限公司 一种新型多环类化合物及其组合物和用途
CN116239566A (zh) * 2021-12-08 2023-06-09 标新生物医药科技(上海)有限公司 E3泛素连接酶配体化合物、基于该配体化合物开发的蛋白降解剂及它们的应用
WO2023151559A1 (fr) * 2022-02-08 2023-08-17 和径医药科技(上海)有限公司 Composé hétérocyclique, composition pharmaceutique le contenant et utilisation anti-tumorale associée
WO2023212599A2 (fr) * 2022-04-26 2023-11-02 Endotarget Inc. Composés et méthodes pour la dégradation ciblée de récepteurs d'œstrogène

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6875775B2 (en) * 1999-11-16 2005-04-05 Hormos Medical Oy Ltd Triphenylalkene derivatives and their use as selective estrogen receptor modulators
WO2013097773A1 (fr) * 2011-12-30 2013-07-04 Centaurus Biopharma Co., Ltd. Nouveaux dérivés d'arylalcène et utilisation de ceux-ci en tant que modulateurs sélectifs de récepteur d'œstrogène
US20150018341A1 (en) * 2011-12-30 2015-01-15 Centaurus Biopharma Co., Ltd. Novel arylalkene derivatives and use thereof as selective estrogen receptor modulators
US20180208590A1 (en) * 2016-07-12 2018-07-26 Accutar Biotechnology Inc. Novel compounds having estrogen receptor alpha degradation activity and uses thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201311891D0 (en) * 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compound
KR102173464B1 (ko) 2016-12-01 2020-11-04 아비나스 오퍼레이션스, 인코포레이티드 에스트로겐 수용체 분해제로서의 테트라히드로나프탈렌 및 테트라히드로이소퀴놀린 유도체
CA3096790C (fr) * 2018-04-09 2024-03-19 Shanghaitech University Compose ciblant une degradation proteique, utilisation antitumorale, intermediaire de celui-ci et utilisation de l'intermediaire
WO2019241231A1 (fr) * 2018-06-11 2019-12-19 Abhishek Sharma Composés anti-oestrogène
CN111205282B (zh) * 2018-11-21 2023-12-01 上海科技大学 Er蛋白调节剂及其应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6875775B2 (en) * 1999-11-16 2005-04-05 Hormos Medical Oy Ltd Triphenylalkene derivatives and their use as selective estrogen receptor modulators
WO2013097773A1 (fr) * 2011-12-30 2013-07-04 Centaurus Biopharma Co., Ltd. Nouveaux dérivés d'arylalcène et utilisation de ceux-ci en tant que modulateurs sélectifs de récepteur d'œstrogène
US20150018341A1 (en) * 2011-12-30 2015-01-15 Centaurus Biopharma Co., Ltd. Novel arylalkene derivatives and use thereof as selective estrogen receptor modulators
US20180208590A1 (en) * 2016-07-12 2018-07-26 Accutar Biotechnology Inc. Novel compounds having estrogen receptor alpha degradation activity and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
International Search Report for International Application No. PCT/US19/62564, dated Mar. 3, 2020. *
Lahoz, et al. Organic Electronics 14 (2013), pp. 1225-1230. *
Written Opinion of the International Search Authority for International Application No. PCT/US19/62564, dated Mar. 3, 2020. *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11642342B2 (en) 2019-12-23 2023-05-09 Accutar Biotechnology Combinations of estrogen receptor degraders and cyclin-dependent kinase inhibitors for treating cancer

Also Published As

Publication number Publication date
EP3675839A4 (fr) 2020-12-23
CN113164409A (zh) 2021-07-23
WO2020106933A1 (fr) 2020-05-28
CN113164409B (zh) 2023-07-18
EP3675839B1 (fr) 2023-03-01
US11117885B2 (en) 2021-09-14
CA3120530A1 (fr) 2020-05-28
US20200299264A1 (en) 2020-09-24
AU2019384807A1 (en) 2021-05-20
US20200157078A1 (en) 2020-05-21
CN117050002A (zh) 2023-11-14
EP3675839A1 (fr) 2020-07-08
SG11202105256QA (en) 2021-06-29

Similar Documents

Publication Publication Date Title
US10696659B2 (en) Compounds having estrogen receptor alpha degradation activity and uses thereof
US10800770B1 (en) Chroman derivatives having estrogen receptor degradation activity and uses thereof
US11220490B2 (en) Ureas having androgen receptor degradation activity and uses thereof
WO2021252339A1 (fr) Composés de purine -2,6-dione substitués en tant qu'inhibiteurs de kras
US20220112204A1 (en) Substituted dihydropyranopyrimidine compounds as kras inhibitors
US10968212B2 (en) Compounds having estrogen receptor alpha degradation activity and uses thereof
US11535606B2 (en) Substituted quinoline-8-carbonitrile derivatives with androgen receptor degradation activity and uses thereof
US20220220124A1 (en) Novel compounds having bet, estrogen receptor, and androgen receptor degradation activity and uses thereof
US10836749B1 (en) Substituted quinoline-8-carbonitrile derivatives having androgen receptor degradation activity and uses thereof
US10501466B2 (en) WDR5 inhibitors and modulators
US11999747B2 (en) Furoquinolinediones as inhibitors of TDP2
US20240190842A1 (en) Substituted quinolinone-8-carbonitrile derivatives having androgen degradation activity and uses thereof
US20240182492A1 (en) Azaheteroaryl compound, preparation method therefor, and application thereof
US20200017485A1 (en) Compounds and methods for regulating insulin secretion

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY